EXOSC3 Human

Exosome Component 3 Human Recombinant
Shipped with Ice Packs
In Stock

Description

EXOSC3 Human Recombinant produced in E.Coli is a single, non-glycosylated polypeptide chain containing 298 amino acids (1-275 a.a) and having a molecular mass of 32.0kDa.
EXOSC3 is fused to a 23 amino acid His-tag at N-terminus & purified by proprietary chromatographic techniques.

Product Specs

Introduction
Exosome Component 3 (EXOSC3) is a vital part of the human exosome complex, which possesses 3'-5' exoribonuclease activity. This component is not directly involved in catalysis but plays a crucial role in various RNA processing and degradation tasks, including the breakdown of histone mRNA. Interestingly, pseudogenes related to EXOSC3 are found on chromosomes 19 and 21.
Description
Recombinant EXOSC3, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 298 amino acids (specifically, amino acids 1-275). It has a molecular weight of 32.0 kDa. For purification purposes, a 23 amino acid His-tag is fused to the N-terminus of EXOSC3. The purification process involves proprietary chromatographic techniques.
Physical Appearance
A clear and sterile solution without any color.
Formulation
The EXOSC3 protein solution has a concentration of 0.5mg/ml and contains 20mM Tris-HCl buffer (with a pH of 7.5), 0.15M NaCl, 30% glycerol, and 1mM DTT.
Stability
For short-term storage (2-4 weeks), the solution can be kept at 4°C. For longer storage, freezing at -20°C is recommended. Adding a carrier protein like HSA or BSA (0.1%) is advisable for long-term storage. Repeated freezing and thawing should be avoided.
Purity
SDS-PAGE analysis confirms a purity greater than 85.0%.
Synonyms
bA3J10.7, CGI-102, hRrp-40, p10, PCH1B, RP11-3J10.8, RRP40, Rrp40p, Exosome complex component RRP40, Exosome component 3, Ribosomal RNA-processing protein 40, EXOSC3.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMAEPASV AAESLAGSRA RAARTVLGQV VLPGEELLLP EQEDAEGPGG AVERPLSLNA RACSRVRVVC GPGLRRCGDR LLVTKCGRLR HKEPGSGSGG GVYWVDSQQK RYVPVKGDHV IGIVTAKSGD IFKVDVGGSE PASLSYLSFE GATKRNRPNV QVGDLIYGQF VVANKDMEPE MVCIDSCGRA NGMGVIGQDG LLFKVTLGLI RKLLAPDCEI IQEVGKLYPL EIVFGMNGRI WVKAKTIQQT LILANILEAC EHMTSDQRKQ IFSRLAES.

Q&A

What is the EXOSC3 gene and what function does it serve in human cells?

The EXOSC3 gene provides instructions for making exosome component 3, a non-catalytic subunit of the RNA exosome complex. This multi-protein complex plays crucial roles in RNA processing and degradation, including cleaving RNA molecules at specific sites and breaking down RNA molecules when they are no longer needed . The RNA exosome processes multiple types of RNA, making it essential for normal cellular functioning. Specifically, EXOSC3 forms part of the cap structure of the exosome complex and is involved in substrate recognition and binding . Proper RNA processing and turnover are fundamental to gene expression regulation and cellular homeostasis across all tissue types.

Where is EXOSC3 expressed in the human body, and are there tissue-specific expression patterns?

EXOSC3 is widely expressed throughout the human body, but it shows particularly important functions in the developing nervous system. Studies suggest that exosome component 3 activity is necessary for normal development and growth of certain brain regions, particularly the cerebellum, which coordinates movement . Additionally, EXOSC3 appears to be essential for the survival of motor neurons in the spinal cord, which are specialized nerve cells that control muscle movement . Tissue expression data from the Allen Brain Atlas indicate varying expression levels across different brain regions, with notable expression patterns in both the adult and developing human brain .

How does the structure of EXOSC3 relate to its function in the RNA exosome complex?

EXOSC3 (also known as RRP40) functions as a non-catalytic component of the human exosome, a complex with 3'-5' exoribonuclease activity . The protein contains specific structural domains that enable it to participate in RNA binding and to interact with other exosome components. While EXOSC3 itself does not possess catalytic activity, its structural features are crucial for the assembly and stability of the entire RNA exosome complex. The protein forms part of the cap structure of the complex, which is essential for substrate recognition and channeling RNA molecules to the catalytic core components. Mutations that affect EXOSC3's structure, particularly those that impair its ability to properly fold or interact with other exosome components, can significantly disrupt RNA processing and lead to disease states.

What types of EXOSC3 mutations have been identified, and how do they correlate with clinical presentations?

At least 16 distinct mutations in the EXOSC3 gene have been identified in patients with pontocerebellar hypoplasia . These mutations result in exosome component 3 proteins with reduced or no function. The most common mutation replaces the amino acid aspartic acid with alanine at protein position 132 (Asp132Ala or D132A) . Interestingly, researchers have observed clear genotype-phenotype correlations, with certain mutations consistently associated with milder phenotypes. For example, patients with the D132A mutation tend to have less severe brain abnormalities compared to those with other EXOSC3 mutations .

The table below summarizes key EXOSC3 mutations and their associated clinical severity:

MutationProtein ChangeClinical SeverityNotable Features
c.395A>CAsp132Ala (D132A)ModerateLess severe brain abnormalities, longer survival
c.92G>CGly31Ala (G31A)SeverePronounced cerebellar hypoplasia, early mortality
c.404G>AArg135His (R135H)VariableSpinal motor neuron involvement
Various deletionsProtein truncationSevereComplete loss of function, profound developmental impairment

What is the molecular pathogenesis of pontocerebellar hypoplasia type 1B (PCH1B) caused by EXOSC3 mutations?

PCH1B results from impaired RNA exosome function due to mutations in the EXOSC3 gene. Approximately half of all cases of pontocerebellar hypoplasia type 1 (PCH1) are caused by EXOSC3 mutations . The molecular pathogenesis involves disruption of essential RNA processing and degradation mechanisms in developing neurons.

While the exact pathways leading from EXOSC3 dysfunction to neuronal death remain under investigation, several mechanisms have been proposed:

  • Accumulation of aberrant RNA species that would normally be degraded by the exosome

  • Defects in ribosomal RNA processing leading to impaired protein synthesis

  • Disruption of specific RNA processing events critical for neuronal development

  • Altered gene expression profiles due to improper mRNA turnover

The selective vulnerability of cerebellar and spinal motor neurons suggests that these cell types may have particularly high requirements for precise RNA metabolism during development . Research indicates that EXOSC3 mutations may cause disease through both loss-of-function mechanisms and potential toxic gain-of-function effects, depending on the specific mutation.

How do genotype-phenotype correlations inform prognosis and disease management in EXOSC3-related disorders?

Genotype-phenotype correlations in EXOSC3-related disorders provide valuable prognostic information. Multiple studies have demonstrated that certain mutations consistently produce milder clinical courses while others result in more severe presentations . For example, patients carrying the D132A mutation typically have a less severe clinical course with somewhat preserved cerebellar development and longer survival compared to those with other mutations .

These correlations have important implications for clinical management, including:

  • Providing more accurate prognostic information to families

  • Guiding decisions about interventional therapies and supportive care

  • Informing surveillance for potential complications

  • Helping to establish appropriate developmental and educational interventions

A comprehensive approach to managing EXOSC3-related disorders requires multidisciplinary care that addresses both neurological symptoms and systemic manifestations. Early identification of the specific EXOSC3 mutation can help clinicians develop more tailored management strategies and better anticipate disease progression.

What are the optimal methods for studying EXOSC3 function in cellular and animal models?

Investigating EXOSC3 function requires a multi-faceted experimental approach:

Cellular Models:

  • CRISPR/Cas9-mediated gene editing to create isogenic cell lines with specific EXOSC3 mutations

  • RNA interference (RNAi) techniques for temporary knockdown studies

  • Overexpression systems with tagged EXOSC3 variants to study protein interactions

  • Patient-derived induced pluripotent stem cells (iPSCs) differentiated into neurons to study disease-relevant phenotypes

Animal Models:

  • Conditional knockout mouse models (complete knockout is often embryonic lethal)

  • Zebrafish models, which offer advantages for studying neurodevelopmental phenotypes

  • Drosophila models for high-throughput genetic interaction studies

Key Analytical Techniques:

  • RNA-seq to assess global changes in RNA processing and abundance

  • CLIP-seq (Cross-linking immunoprecipitation followed by sequencing) to identify direct RNA targets

  • Mass spectrometry to characterize protein interaction partners

  • Immunohistochemistry to examine tissue-specific expression patterns

When designing experiments, it's crucial to consider the developmental timing of EXOSC3 expression, as its effects on neurodevelopment may occur during specific embryonic windows. Additionally, tissue-specific conditional knockout models can help distinguish between developmental and maintenance roles of EXOSC3 in different neuronal populations.

What methodologies are most effective for analyzing RNA processing defects in EXOSC3-mutant cells?

Analyzing RNA processing defects in EXOSC3-mutant cells requires sophisticated molecular biology techniques:

  • Global RNA analysis:

    • RNA-seq with specialized library preparation to capture various RNA species (mRNA, rRNA, snoRNAs)

    • Northern blotting to detect specific RNA processing intermediates

    • Quantitative RT-PCR for targeted analysis of known exosome substrates

  • RNA stability assays:

    • Actinomycin D chase experiments to measure RNA half-lives

    • Metabolic labeling with 4-thiouridine followed by sequencing (4sU-seq) to measure RNA synthesis and decay rates

    • BRIC (5′-bromo-uridine immunoprecipitation chase) to track newly synthesized RNA

  • RNA structure and interaction analysis:

    • SHAPE (Selective 2′-hydroxyl acylation analyzed by primer extension) to examine RNA structural changes

    • RIP-seq (RNA immunoprecipitation sequencing) to identify RNAs bound by exosome components

    • Proximity labeling approaches to characterize the RNA exosome interactome

  • Single-cell approaches:

    • Single-cell RNA-seq to detect cell-type-specific effects

    • RNA FISH (fluorescence in situ hybridization) to visualize RNA localization and abundance at the single-cell level

When analyzing results, it's important to distinguish primary effects (direct consequences of EXOSC3 dysfunction) from secondary adaptations. Comparing multiple model systems and mutation types can help identify consistent molecular signatures of exosome dysfunction.

How can high-throughput screening approaches be applied to identify potential therapeutic targets for EXOSC3-related disorders?

High-throughput screening offers promising avenues for identifying therapeutic targets for EXOSC3-related disorders:

  • CRISPR screens:

    • Genome-wide CRISPR knockout or activation screens to identify genetic modifiers of EXOSC3 phenotypes

    • Focused CRISPR libraries targeting RNA processing pathways to find synthetic lethal or rescue interactions

  • Small molecule screens:

    • Cell-based phenotypic screens using patient-derived cells or engineered reporter lines

    • Target-based screens focusing on EXOSC3 protein stability or interactions

    • RNA-targeted small molecule screens to identify compounds that can correct specific RNA processing defects

  • Transcriptome-based approaches:

    • Analysis of differential gene expression patterns to identify compensatory pathways

    • Connectivity mapping to find drugs that reverse the transcriptional signature of EXOSC3 deficiency

  • Therapeutic modality screening:

    • Antisense oligonucleotides to modify specific RNA processing events

    • mRNA therapeutics to transiently supplement EXOSC3 function

    • Protein stabilization approaches for missense mutations that affect protein stability

When implementing high-throughput screens, careful selection of assay endpoints is critical. For EXOSC3-related disorders, relevant endpoints might include markers of neuronal differentiation, RNA exosome assembly or activity, or specific RNA processing events known to be disrupted in patient cells. Validation studies should include multiple cell types relevant to the disease, particularly cerebellar neurons and motor neurons.

How does EXOSC3 selectivity for RNA substrates contribute to tissue-specific phenotypes in disease states?

The tissue-specific phenotypes observed in EXOSC3-related disorders, particularly the vulnerability of cerebellar and motor neurons, raise intriguing questions about substrate selectivity:

The RNA exosome processes numerous RNA species, yet mutations in EXOSC3 predominantly affect specific neuronal populations. This selectivity likely stems from several factors:

  • Tissue-specific RNA expression:

    • Certain RNA species that rely heavily on exosome processing may be predominantly expressed in vulnerable neuronal populations

    • Cell-type-specific alternative splicing events may generate transcripts that are particularly dependent on exosome function

  • Developmental timing:

    • Cerebellar and motor neuron development occurs during specific embryonic and early postnatal periods when precise RNA quality control is critical

    • Temporal regulation of RNA processing may be particularly important during neuronal differentiation and maturation

  • Compensatory mechanisms:

    • Different tissues may have varying abilities to compensate for partial exosome dysfunction

    • Alternative RNA degradation pathways may be more or less active across different cell types

Recent research using techniques like CLIP-seq has begun to identify cell-type-specific RNA targets of the exosome complex. Studies in model organisms suggest that defects in processing specific microRNAs and long non-coding RNAs may contribute to neuronal phenotypes. Further research using single-cell transcriptomics and spatial transcriptomics will be essential to fully understand the basis of selective neuronal vulnerability in EXOSC3-related disorders.

What is the role of EXOSC3 in non-coding RNA metabolism and how might this contribute to disease pathogenesis?

EXOSC3, as part of the RNA exosome complex, plays critical roles in the metabolism of numerous non-coding RNA species:

  • rRNA processing:

    • The exosome is essential for proper maturation of ribosomal RNAs

    • EXOSC3 mutations may affect rRNA processing, potentially leading to nucleolar stress and p53 activation

    • Ribosome biogenesis defects could particularly impact highly metabolically active neurons

  • snoRNA and snRNA regulation:

    • Small nucleolar RNAs (snoRNAs) and small nuclear RNAs (snRNAs) are processed and degraded by the exosome

    • Disruption of these processes can affect RNA modifications and splicing patterns

    • Altered RNA modification landscapes may have widespread effects on translation efficiency

  • lncRNA turnover:

    • Long non-coding RNAs often have regulatory functions in neurodevelopment

    • Improper degradation of lncRNAs may disrupt gene expression programming during critical developmental windows

  • miRNA regulation:

    • The exosome may regulate the availability of certain microRNA precursors

    • Changes in miRNA profiles could affect neuronal differentiation and maintenance

The pathogenesis of EXOSC3-related disorders likely involves disruption of multiple RNA processing events rather than a single RNA species. Advanced transcriptomic analyses of patient samples have revealed complex patterns of both increased and decreased abundance of various non-coding RNAs. Understanding these complex RNA networks will be essential for developing targeted therapeutic approaches.

What are the latest findings on the structural biology of EXOSC3 and how can this information guide drug discovery efforts?

Recent advances in structural biology have provided important insights into EXOSC3's role within the RNA exosome complex:

Cryo-electron microscopy and X-ray crystallography studies have revealed that EXOSC3 forms part of the cap structure of the exosome, where it participates in substrate recognition and binding. The three-dimensional structure shows how EXOSC3 interacts with other cap proteins (EXOSC1 and EXOSC2) and with the core ring structure of the exosome.

Key structural insights with therapeutic implications include:

  • RNA binding channels:

    • EXOSC3 contributes to the formation of RNA binding channels that guide substrates to the catalytic core

    • These channels represent potential sites for small molecule modulation of exosome function

  • Protein-protein interaction surfaces:

    • Specific interfaces between EXOSC3 and other exosome components could be targeted to stabilize the complex

    • Small molecules that enhance these interactions might partially restore function of certain mutant EXOSC3 proteins

  • Allosteric regulation sites:

    • The exosome complex undergoes conformational changes during RNA processing

    • Molecules that bind to allosteric sites could potentially enhance residual activity in mutant complexes

  • Mutation-specific structural effects:

    • Mapping disease-causing mutations onto the structure reveals different structural consequences

    • Some mutations affect protein stability while others disrupt specific interactions

Drug discovery efforts based on these structural insights are focusing on several approaches:

  • Protein stabilizers that might rescue folding-defective mutants

  • PROTACs (proteolysis targeting chimeras) to selectively degrade dysfunctional protein complexes

  • RNA mimetics that could help bypass defective exosome function

  • Small molecules that modulate interactions with regulatory proteins

These structure-based approaches represent promising avenues for developing the first targeted therapies for EXOSC3-related disorders.

What are the most sensitive and specific diagnostic approaches for identifying EXOSC3-related disorders?

Diagnosing EXOSC3-related disorders requires a comprehensive approach combining clinical, imaging, and molecular assessments:

Clinical Diagnostic Pathway:

  • Clinical Presentation:

    • Developmental delay and regression

    • Microcephaly (unusually small head size)

    • Intellectual disability

    • Motor problems related to anterior horn cell degeneration

    • Cerebellar signs (ataxia, nystagmus)

  • Neuroimaging:

    • MRI showing pontocerebellar hypoplasia (underdevelopment of the pons and cerebellum)

    • Progressive cerebellar atrophy on sequential imaging

    • Possible spinal cord atrophy

  • Electrophysiological Studies:

    • Electromyography (EMG) showing denervation consistent with anterior horn cell disease

    • Nerve conduction studies to distinguish from peripheral neuropathies

  • Molecular Testing:

    • Targeted EXOSC3 sequencing as first-line genetic testing for patients with PCH1 features

    • Next-generation sequencing panels for pontocerebellar hypoplasia/neurodegeneration

    • Whole exome or genome sequencing for atypical cases

    • RNA sequencing to detect splicing mutations or large deletions that might be missed by exome sequencing

  • Biochemical Assays:

    • Functional assessment of RNA exosome activity in patient-derived cells

    • Analysis of specific RNA processing events known to be affected by EXOSC3 dysfunction

Early and accurate diagnosis is crucial for appropriate management, genetic counseling, and potential enrollment in clinical trials. The combination of characteristic clinical and radiological features with molecular confirmation provides the most reliable diagnostic approach.

How can emerging therapeutic approaches be applied to EXOSC3-related neurological disorders?

Several emerging therapeutic approaches show promise for EXOSC3-related disorders:

  • Gene Therapy Approaches:

    • AAV-mediated gene replacement therapy targeting the CNS

    • Challenges include achieving adequate distribution in the cerebellum and spinal cord

    • Ongoing preclinical studies in animal models show promise for restoring EXOSC3 function

  • RNA-based Therapeutics:

    • Antisense oligonucleotides to modulate splicing or increase expression of compensatory genes

    • mRNA delivery systems to transiently supplement EXOSC3 function

    • Small interfering RNAs to downregulate negative regulators of RNA processing

  • Small Molecule Approaches:

    • Protein stabilizers for missense mutations that affect protein stability

    • Compounds that enhance residual exosome activity

    • Drugs targeting downstream pathways activated by exosome dysfunction

  • Cell-based Therapies:

    • Neural stem cell transplantation to replace affected neurons

    • Particularly challenging due to the widespread nature of the disease

  • Combination Approaches:

    • Targeting multiple aspects of disease pathogenesis simultaneously

    • Combining neuroprotective strategies with approaches to restore EXOSC3 function

The development of these therapies faces several challenges, including the early developmental role of EXOSC3, the need to target both the cerebellum and spinal cord, and the complexity of RNA processing pathways. Clinical trial design for these rare disorders will require innovative approaches to outcome measures and patient stratification based on genotype.

What insights can systems biology approaches provide into compensatory mechanisms that might be leveraged for therapeutic intervention?

Systems biology approaches have revealed potential compensatory mechanisms that could be therapeutically exploited:

  • Parallel RNA Processing Pathways:

    • Alternative exoribonucleases may partially compensate for exosome dysfunction

    • Upregulation of these pathways through small molecules or genetic approaches could alleviate RNA processing defects

  • Stress Response Pathways:

    • Cells with EXOSC3 mutations show activation of various stress response pathways

    • Targeted modulation of these responses might enhance neuronal survival

  • Translational Reprogramming:

    • Changes in translation efficiency may help cells adapt to altered RNA processing

    • Compounds that enhance specific aspects of translational control could be beneficial

  • Metabolic Adaptations:

    • Neurons with compromised RNA processing show altered metabolic profiles

    • Metabolic interventions might support neuronal function despite ongoing RNA processing defects

Multi-omics studies (combining transcriptomics, proteomics, and metabolomics) in patient-derived cells have identified networks of genes that respond to EXOSC3 dysfunction. Computational analyses have highlighted potential hub genes that might serve as therapeutic targets. For example, certain RNA binding proteins appear to be upregulated in response to exosome dysfunction and may partially compensate by stabilizing critical transcripts.

These systems-level insights suggest that therapeutic approaches might focus not only on restoring EXOSC3 function but also on enhancing compensatory mechanisms that naturally emerge in response to exosome dysfunction. Drug repurposing screens guided by these network analyses have identified several FDA-approved compounds that appear to enhance these compensatory responses and are now being evaluated in preclinical models.

Product Science Overview

Structure and Composition

EXOSC3 is a single, non-glycosylated polypeptide chain containing 298 amino acids, with a molecular mass of approximately 32.0 kDa . The recombinant form of EXOSC3 is typically produced in Escherichia coli and is fused to a 23 amino acid His-tag at the N-terminus for purification purposes .

Function

The primary role of EXOSC3 within the exosome complex is to assist in the processing and degradation of various RNA molecules. This activity is essential for the normal functioning of cells, as it ensures the appropriate processing and breakdown of RNA molecules when they are no longer needed . EXOSC3 is particularly important for the degradation of histone mRNA, which is a critical process during cell division .

Biological Significance

EXOSC3 is necessary for the normal development and growth of certain areas of the brain, particularly the cerebellum, which coordinates movement. It is also vital for the survival of specialized nerve cells in the spinal cord called motor neurons, which play an essential role in muscle movement .

Health Implications

Mutations in the EXOSC3 gene have been linked to a disorder of brain development known as pontocerebellar hypoplasia type 1B (PCH1B). This condition is characterized by delayed development, microcephaly (an unusually small head size), intellectual disability, and problems with muscle movement due to the loss of motor neurons in the spinal cord . The most common mutation involves the replacement of the amino acid aspartic acid with alanine at position 132 (Asp132Ala or D132A), leading to a protein with reduced or no function .

Production and Storage

Recombinant EXOSC3 is produced in Escherichia coli and is available as a sterile, filtered, colorless solution. It is formulated in a buffer containing 20 mM Tris-HCl (pH 7.5), 0.15 M NaCl, 30% glycerol, and 1 mM DTT . For long-term storage, it is recommended to store the protein at -20°C with a carrier protein to avoid multiple freeze-thaw cycles .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.